Protein Micro Arrays

Embed Size (px)

Citation preview

  • 7/31/2019 Protein Micro Arrays

    1/10

    REVIEW

    Protein microarrays for diagnostic assays

    Michael Hartmann & Johan Roeraade & Dieter Stoll &Markus F. Templin & Thomas O. Joos

    Received: 24 June 2008 /Revised: 6 August 2008 /Accepted: 1 September 2008 /Published online: 20 September 2008# Springer-Verlag 2008

    Abstract Protein microarray technology has enormous po-

    tential for in vitro diagnostics (IVD). Miniaturized parallelizedimmunoassays are perfectly suited to generating a maximum

    of diagnostically relevant information from minute amounts

    of sample whilst only requiring small amounts of reagent.

    Protein microarrays have become well-established research

    tools in basic and applied research and the first products are

    already on the market. This article reviews the current state of

    protein microarrays and discusses developments and future

    demands relating to protein arrays in their role as multiplexed

    immunoassays in the field of diagnostics.

    Keywords Protein microarrays . Multiplexed diagnostics .

    In-vitro diagnostics . Focused protein-profiling .

    Analytical microarrays

    Introduction

    Ambient analyte theory

    Immunoassays have been widely employed as highly sensi-

    tive tools for almost half a century [1, 2]. Antibody-based

    immunoassays enable the generation of highly robust assays

    that can be easily standardized and automated. Nowadays

    there are hundreds of antibody-based assays on the diagnos-

    tic market [3]. The miniaturization of immunoassays fordiagnostic purposes started way back in the early sixties [4,

    5]. Feinberg and his colleagues developed a microspot-based

    assay that enabled them to diagnose autoimmune diseases.

    Auto-antigens were immobilized in a microspot and incu-

    bated with human serum. The presence of auto-antibodies in

    the serum led to the spontaneous precipitation of the auto-

    antigens in the microspot. The authors assumed that such

    microspot assays would have a particular advantage for

    routine use on clinical specimens because it is simple,

    sensitive, objective, quickly carried out and read, requires but

    minute quantities of serum and antigen, and provides a

    permanent record for the case files. However, this micro-

    spot-based assay format did not have many supporters. More

    than two decades later, Roger Ekins came up with the

    ambient analyte theory on the feasibility of highly sensitive

    multi-spot and multi-analyte immunoassays [6]. According

    to Ekins theory, miniaturization also leads to an increase in

    detection sensitivity1. According to the law of mass action,

    only a small amount of analyte molecules will be captured on

    to a spot that only contains a minute amount of immobilized

    capture molecules. Under ambient analyte conditions, the

    concentration of analyte in the sample will not change

    significantly even though the capture molecule is a high-

    affinity binder and the analyte concentration is low (Fig. 1).

    Therefore, this type of miniature immunoassay is concentra-

    tion-dependent: the analyte molecules captured in the spot

    directly reflect the analyte concentration in the sample. In

    consequence of the unaltered analyte concentration, the signal

    becomes independent of the sample volume. High sensitivity

    Anal Bioanal Chem (2009) 393:14071416

    DOI 10.1007/s00216-008-2379-z

    M. Hartmann : J. Roeraade

    School of Chemical Science and Engineering,

    Department of Analytical Chemistry,

    Royal Institute of Technology,

    10044 Stockholm, Sweden

    M. Hartmann : D. Stoll : M. F. Templin : T. O. Joos (*)

    NMI Natural and Medical Sciences

    Institute at the University of Tbingen,

    72770 Reutlingen, Germany

    e-mail: [email protected]

    1 Here detection sensitivity is used in terms of limit of detection, i.e.

    the smallest amount of analyte which can be detected with acceptable

    statistical significance. By contrast we use the term assay sensitivity,

    i.e. the smallest detectable difference in analyte concentration.

  • 7/31/2019 Protein Micro Arrays

    2/10

    is achieved because the measurement always takes place at the

    highest analyte concentration possible. Thus, for a given

    analyte concentration, it is possible that the detection

    sensitivity in a microspot is higher than in a macrospot,

    because the analyte is contained in a small area and only signaldensity (i.e. signal per area) not the total sum of signal is

    relevant for signal-to-noise ratios defining the lower limit of

    detection. For this reason and the possibility of being able to

    detect multiple analytes in a single experiment, Ekins was

    convinced that miniaturized and parallelized protein micro-

    arrays had enormous potential for diagnostic applications [6].

    From genomics to proteomics

    Despite protein microarrays huge potential for diagnostic

    applications, it was, nevertheless, the field of genomics that

    made the greatest contribution to the advance of microarray

    technology. Following the sequencing of the entire human

    genome, DNA microarrays were developed and applied to

    large-scale genomics research. Comprising up to tens of

    thousands of different oligonucleotide probes per square

    centimeter, DNA microarrays are perfectly suited to high-

    throughput hybridization systems that enable the expression

    analysis of the entire transcriptome in a single experiment

    [7, 8]. Nowadays, DNA microarrays are well-established

    and reliable methods for mRNA expression profiling and

    SNP analysis. In addition, the first diagnostic assays have

    been placed on the market [9, 10]. Despite the huge potential

    of DNA, it is nevertheless proteins that are the key players in

    cellular processes. DNA microarrays only provide a limited

    amount of information on the status of cells and tissues.

    Cellular functions depend on protein activity, and proteins

    are not only regulated by the differential expression of the

    underlying genes but also by post-translational modifica-tions. Furthermore, protein expression does not generally

    quantitatively correlate with mRNA expression [11]. Within

    the last decade, protein microarrays have entered the field

    of proteomic research [12, 13] demonstrating their huge

    capacity for identifying and quantifying proteins and for

    studying the function of proteins from the perspective of

    the proteome as a whole [14]. Besides their application

    in proteome-wide research and the determination of the

    biochemical activities of proteins, protein microarrays are

    also perfect tools for quantitating subsets of proteins in com-

    plex mixtures. This approach is known as focused protein-

    profiling or analytical microarrays. As correctly foreseen byRoger Ekins, multiplex immunoassays like these have been

    adapted to the microarray format and are about to be used in

    diagnostic applications. This article reviews the present state

    of multiplex immunoassays and discusses the obstacles that

    still slightly hamper their breakthrough in IVD.

    Protein microarrays

    Principles and basics

    DNA microarrays preceded the use of protein microarrays.

    When proteins rather than DNA are used, a variety of

    additional challenges arise due to the more complex nature

    of proteins. A DNA molecule is built up from four different

    bases that all have the same hydrophilic sugar backbone.

    DNA is a very uniform and stable molecule due to its

    chemical structure and the pairing of complementary bases.

    DNA molecules exhibit a strong one-to-one interaction,

    which, under physiological conditions, is biochemically

    more or less inert. Proteins, which are a lot more fragile, are

    assembled from 20 different amino acids that are extremely

    heterogeneous in size and can have either hydrophobic or

    hydrophilic side chains. The proper functioning of proteins

    depends on their tertiary and quaternary structures. The

    foundation of these structures is a balanced system of

    electrostatic forces, hydrogen bonds, and hydrophobic Van

    der Waals interactions. In contrast with DNA, slight changes

    in salt concentration or pH, the presence of oxidants, or the

    removal of water often irreversibly harms their structure and

    hence interferes with their function. Furthermore, PCR and the

    ability to chemically synthesize oligonucleotides made a con-

    siderable contribution to the success of DNA microarray

    analytics. The amplification of proteins is impossible. High-

    Fig. 1 Signal and signal density in microspots. Signal density (signal/

    area) and signal (total intensity) of captured targets in microspots are

    shown for different concentrations of capture molecules. The capture

    molecules are immobilized with the same surface density on all spots.

    The signal (total signal) increases with increasing amount of capture

    molecules for growing spot size. When most of the targets are capturedfrom the solution the signal reaches its maximum. By contrast, signal

    density (signal/area) increases with decreasing amount of capture

    molecules (decreasing spot size), reaching a constant level when the

    capture molecule concentration is

  • 7/31/2019 Protein Micro Arrays

    3/10

    affinity and high-specificity capture molecules can be easily

    predicted and generated from the primary sequence of the

    target DNA. Protein capture molecules cannot be predicted or

    designed on the basis of a similarly easy principle, because the

    interaction underlies a broad range of the aforementioned

    molecular forces. In addition, post-translational modifications

    such as phosphorylation, acetylation, or glycosylation have to

    be taken into account when designing capture or bindingmolecules. At present, the lack of highly specific and high-

    affinity capture molecules is still the main limitation of protein

    microarrays.

    Antibodies are broadly used in research and diagnostic

    applications because of their ability to bind to target

    proteins with high specificity and high affinity. When

    protein microarrays were still in their infancy, scientists

    initially resorted to antibodies since these proteins were

    well known from immunoassays [1]. The invention of

    monoclonal antibodies was a major step forward in the

    generation of unlimited resources of defined capture

    molecules. From then on, it was possible to produce pureand highly specific antibodies against almost any type of

    antigen [15]. However, the generation and validation of

    antibodies, regardless of whether they are monoclonal or

    polyclonal, is a time-intensive and cost-intensive process.

    In-vitro strategies that enabled the generation of binding

    molecules were eventually developed. For instance, phage-

    display technology can be used to screen large synthetic

    libraries of protein clones, within a few weeks, for detection

    of suitable binders against a target molecule of interest [16].

    However, it also turned out that additional maturation steps

    were needed for generation of high-affinity binders that

    were similar to monoclonal or polyclonal antibodies.

    Another promising strategy for producing synthetic binders

    is the generation of aptamers. Aptamers are short single-

    stranded nucleic acid oligomers (ssDNA or RNA) with a

    specific and complex three-dimensional shape which causes

    their well-fitting binding. Aptamers are produced using an

    in-vitro selection and amplification technique called

    SELEX (systematic evolution of ligands by exponential

    enrichment). Many of the selected aptamers show affinities

    comparable with those observed for monoclonal antibodies

    [17]. Affibodies are another class of binding molecule.

    They are based on combinatorial protein engineering of the

    small and robust -helical structure of the domains of

    protein A [18, 19]. These synthetic binders are more robust

    than antibodies, which makes them perfectly suitable as

    capture or detection agents in protein microarrays. However,

    they had to prove that they had an affinity and specificity that

    was similar to that of antibodies. Despite all the advances in

    recombinant and scaffold-based technologies, the most

    advanced binding reagent project today, the Human Protein

    Atlas, uses a polyclonal antibody approach to generate

    binding reagents against human proteins [20].

    Planar microarrays

    Protein microarrays are highly miniaturized and parallelized

    solid-phase assay systems that use a large number of

    different capture molecules immobilized in microspots

    (diameter

  • 7/31/2019 Protein Micro Arrays

    4/10

    based systems are similar to those obtained with established

    ELISA systems involving planar arrays. Some bead-basedimmunoassays proved to be as sensitive, accurate, and

    precise as competitive radio immunoassays [26]. Nowa-

    days, the most popular platform is Luminexs xMAP

    technology. xMAP differentiates 100 different color-coded

    beads and allows researchers to easily set-up multiplex

    assays with small numbers of analytes; alternatively, they

    can choose from the increasing number of commercially

    available kits (http://www.luminexcorp.com ). The BD

    FACSArray Bioanalyzer (http://www.bdbiosciences.com )

    or standard FACS instruments are able to classify the

    different bead types by their size and can use two or more

    different excitation lasers for multiplexed detection. This

    allows the design of more complex assays. At present, the

    washing steps in bead-based assays mainly involve filter

    plates. However, the implementation of magnetic beads will

    further simplify automation; problems experienced with

    filter plates such as clogging, leaking, or unspecific

    adsorption of analyte on to the large surface of filters will

    be avoided. The integration of established magnetic bead-

    handling technologies such as magnetic plate separators

    [27], magnetic pin heads (www.thermo.com/kingfisher), or

    in-tip magnetic capture (http://www.magbio.com) are a

    decisive step towards the further automation of bead arrays.

    Assay formats

    Besides protein expression analysis, protein microarrays

    can also be used for the functional analysis of proteins,

    including protein interaction involving immobilized pro-teins or peptides, low molecular weight compounds, DNA,

    oligosaccharides, tissues, or cells. In general, protein array

    assay formats can be divided into forward-phase and

    reversed-phase arrays (Fig. 3). Forward-phase protein

    microarray assays involve the immobilization of capture

    agents and hence enable the analysis of multiple parameters

    from a single sample that is incubated on the array. In

    reversed-phase protein microarrays, many different samples

    (cell or tissue lysates) are immobilized in a microarray

    format and are simultaneously analyzed for the presence of

    a single target protein using a target-specific antibody.

    Replicates of such protein arrays allow the analysis ofhundreds of parameters from minimal amounts of sample.

    The reversed-phase array format is ideally suited to looking

    into large sample cohorts. It enables the detection of

    differentially regulated proteins in healthy or diseased

    tissue and treated and untreated cells, the identification of

    disease-specific biomarkers, or the analysis of cell signaling

    networks [28]. Tissue arrays are a special type of reversed-

    phase microarray and consist of tissue slices that are

    immobilized on a surface. They are the miniature equiva-

    lent of classical immunohistochemistry assays and enable

    Fig. 2 Planar and bead-based microarrays. In planar microarrays,

    individual capture agents are immobilized in a microarray format

    containing between several hundreds to several thousands of spots.

    The arrays are probed with sample and the analytes of interest bind to

    their cognate capture agent. The binding reaction is verified by a

    fluorescence read out. In bead-based microarrays, individual capture

    agents are bound to color-coded or size-coded microspheres. The

    assay can be performed with standard laboratory ware; a flow

    cytometer is used to detect the fluorescent label

    Fig. 3 Forward and reversed-phase array format. In a forward-phase

    array format, a large number of immobilized capture molecules enable

    the analysis of many parameters from a single sample, regardless

    whether the sample is directly labeled or a sandwich assay is

    performed. In a reversed-phase array, many samples are immobilized

    and a highly specific antibody used to analyze the expression of a

    single parameter in the immobilized samples

    1410 M. Hartmann et al.

    http://www.luminexcorp.com/http://www.bdbiosciences.com/http://www.thermo.com/kingfisherhttp://www.magbio.com/http://www.magbio.com/http://www.thermo.com/kingfisherhttp://www.bdbiosciences.com/http://www.luminexcorp.com/
  • 7/31/2019 Protein Micro Arrays

    5/10

    the simultaneous analysis of large numbers of tissues with

    minimum reagent consumption [29].

    Currently, forward-phase assays are the most frequently

    used protein microarray format. Protein arrays are used for

    detailed expression analysis and for functional protein

    studies. Forward-phase protein microarray assays are

    mainly applied for antibody arrays to quantify dozens of

    analytes that are present in complex samples. In antibodyarrays, antibodies are immobilized on a carrier at densities

    of a few up to several thousand antibodies. Captured

    analytes can either be visualized by directly labeling the

    samples (analogous to DNA microarrays) or by multiplexed

    sandwich immunoassays. Protein microarrays enable

    screening for disease-related up or down-regulation of

    proteins in patient samples [30]. Fluorescence labeling of

    a sample necessitates careful optimization in order to obtain

    optimum signal-to-noise ratios. Insufficient labeling proce-

    dures result in a decrease in detection sensitivity whereas

    over-labeling of a sample will cause high background

    signals. The sample can also be labeled with biotin andsubsequently incubated with fluorescence-labeled streptavi-

    din; this might improve signal-to-noise ratios [31]. How-

    ever, it has to be kept in mind that the labeling of the target

    proteins may interfere with the antibody-antigen interac-

    tion, because the labeling procedure could negatively affect

    the epitope and destroy it. Finally, one has to be aware that

    proteins appear in complexes. A strong signal can thus

    either be derived from large amounts of target analytes

    captured or, alternatively, from capturing a huge protein

    complex. The use of a single capture reagent cannot

    discriminate between a specific and an unspecific binding

    event. Specificity is greatly enhanced by using matched

    antibody pairs in miniaturized sandwich immunoassays in

    which high-affinity antibodies are immobilized in an array

    format and capture the target analytes during incubation

    with the sample. Unbound molecules are washed off

    and the array is incubated with detection antibodies that

    bind to another epitope on the target analytes. In sand-

    wich immunoassays, two separate binding reactions are

    employed; this is substantially more specific than direct

    labeling approaches. The detection antibody can be directly

    labeled with fluorescence; alternatively, the signal can be

    further enhanced when using the biotin-streptavidin re-

    porter system. The biotin-streptavidin model is a universal

    reporter system that simplifies the assay procedure, because

    all detection antibodies can be used in a biotinylated form.

    Despite high selectivity and detection sensitivity, it is,

    nevertheless, not possible to achieve unlimited multiplexing

    of sandwich immunoassays due to cross reactivity arising

    from the increasing overall concentration of the cocktail of

    different detection antibodies, which increases background

    signals above a certain threshold. In all complex multi-

    plexed sandwich immunoassays discussed so far, multi-

    plexed sandwich immunoassays used for the detection of

    up to 100 analytes had to be divided into several multi-

    plexed assays [32]. In an 11-plex sandwich immunoassay,

    the detection sensitivity decreased by a factor of 1.7up to

    5 compared with the single-plex assays, because higher

    background signals wer e observ ed in the multip lex

    format [33].

    Another forward-phase assay type uses recombinantlyexpressed proteins that are immobilized as capture mole-

    cules in a microspot. For example, in a study on protein

    interaction, Zhu et al. arrayed 5,800 recombinant yeast

    proteins on a microscope slide and probed this yeast

    proteome microarray with proteins and phospholipids to

    identify new interaction partners [14]. They confirmed the

    already known existence of calmodulin-interacting and

    phospholipid-interacting proteins and identified new inter-

    acting proteins. These types of protein microarray can also

    be used as antigen arrays to study the cross reactivity of

    antibodies or to screen for autoantibodies against unknown

    types of antigen. They represent a special forward-phaseassay type, because the immobilized antigens are not real

    capture agents; instead the antibodies under analysis bind to

    the immobilized antigens. Antigen microarrays are easy to

    establish, because only one species-specific detection anti-

    body is needed to identify bound antibodies in the microspots.

    Antigen microarrays are ideally suited to the multiparametric

    challenges of autoimmune and allergy diagnostics. Several

    autoimmune and allergy microarray assays are already

    commercially available and will be discussed in more detail

    in the next section.

    Other analytical tools

    Apart from the above mentioned classical microarray

    methods, other technologies are employed to analyze

    protein interactions in a multiplex fashion. Very interesting

    are label-free detection methods, because there is no need

    for extra detection agents. For example, Quadraspecs

    spinning disc interferometry (SDI) technology enables

    detection of up to 128 unique analytes in 264 samples per

    disk. They set up a veterinary diagnostic test for canine

    heartworm (www.quadraspec.com). Analytik Jena com-

    bined reflectometric interference spectroscopy (RIfS) with

    biochip technology in their BIAffinity (www.analytik-jena.

    de). Maven Biotechnologies (www.mavenbiotech.com)

    LFIRE is an imaging system based on total internal

    reflection ellipsometry that measures molecular binding

    reactions in a microarray. According to the company,

    LFIRE has been validated with protein microarray densities

    of 2,500 spots cm2 and is capable of detecting molecules

    as small as 150 Daltons. Finally, well established surface

    plasmon resonance (SPR) technology has been adopted to

    microarrays: in research applications, the Biacore Flexchip

    Protein microarrays for diagnostic assays 1411

    http://www.quadraspec.com/http://www.analytik-jena.de/http://www.analytik-jena.de/http://www.mavenbiotech.com/http://www.mavenbiotech.com/http://www.analytik-jena.de/http://www.analytik-jena.de/http://www.quadraspec.com/
  • 7/31/2019 Protein Micro Arrays

    6/10

    has been used to detect up to 400 reactions in parallel [ 34,

    35]. Apart from optical devices there are also electrochem-

    ical platforms, for example the GRAVI-Chip from Diag-

    noSwiss that uses electrochemical read out of enzymatic

    reactions to detect a current that is proportionate to the

    analyte concentration. Basically, many label-free technolo-

    gies allow real-time detection of analytes and, therefore, the

    analysis of kinetic parameters of analytes could giveadditional diagnostic information. However, label-free

    technologies have to prove to be reliable enough to enter

    the market of multiplexed applications.

    This has already been proven by the Triage system (www.

    biosite.com) and the VIDAS platform (www.biomerieux-

    usa.com). Both are micro-fluidic and integrated devices

    based on fluorescent read out that offer a set of multiplex

    assays for detection of proteins related to cardiac diseases

    (Table 1). The development of technology is also driven by

    reducing costs for diagnostics. For example, common

    recordable compact disks as molecular screening surfaces

    and a standard optical CD/DVD drive as detector, havebeen reported [36, 37]. Alpha-fetoprotein has been detected

    in a buffer environment by enzyme or gold nanoparticle-

    labeled antibodies that were used as tracers, forming a

    precipitate on the sensing disk surface. Such low-cost devices

    could also contribute to a better acceptance of microarrays in

    IvD.

    Protein microarrays for diagnostics

    Analytical protein microarrays for detection of antibodies

    A very early example of a multiplex assay system is the

    MASTpette test chamber which was developed for multi-

    parametric allergy testing in the 1980s [38]. Allergen-coated

    cellulose threads were bonded in the test chamber andincubated with patient serum. Bound IgEs were detected

    with enzyme-labeled anti IgE antibody using a chemilumi-

    nescent reaction. Today, IVD MASTpette test chamber-

    based test systems use as little as 230 L for testing 20

    allergen reactions in one assay (www.invernessmedical.de).

    The multiplex detection of allergens was transferred to

    protein microarrays, which has led to further miniaturiza-

    tion and parallelization. One of the first protein microarrays

    for detection of IgE used crude allergen extracts that were

    immobilized in microspots on modified glass slides. This

    approach involved rolling circle amplification in order to

    achieve sufficient sensitivity [39]. Hiller et al. [40] used 94purified allergen molecules representing the most common

    allergen sources to generate an allergen array to screen for

    specific IgEs from minimal amounts of plasma samples.

    Purified allergens enabled the scientists to detect bound IgEs

    without needing to amplify them. Nowadays, protein micro-

    arrays enable the detection of allergen-specific IgE reactivity

    Table 1 Commercial multiplexed and protein-based diagnostics

    Indication Target Vendor Platform FDA cleared

    Allergies Antibodies VBC-Genomics (www.vbc-genomics.at) Planar array

    Allergies, celiac disease Antibodies INOVA Diagnostics (www.inovadx.com) Luminex +

    Allergies, common Antibodies ImmuneTech (www.immunetech.com) Luminex +

    Allergies, indoor allergens Antibodies INDOOR Biotechnologies (www.inbio.com) Luminex

    Autoimmune Antibodies BioArray Solutions (www.bioarrays.com) Bead array +

    Autoimmune Antibodies Biomedical Diagnostics (www.bmd-net.com) Luminex +

    Autoimmune Antibodies INOVA Diagnostics (www.inovadx.com) Luminex +

    Autoimmune Antibodies Zeus Scientific (www.zeusscientific.com) Luminex +

    Autoimmune Antibodies Bio-Rad Laboratories (www.bio-rad.com ) Luminex +

    Autoimmune Antibodies Whatman (www.whatman.com) Planar array

    Cancer Proteins RBM (www.rulesbasedmedicine.com) Luminex

    Cardiac, heart failure Proteins Biomrieux (www.biomerieux-diagnostics.com) VIDAS +

    Cardiac, myocardial infarction Proteins Biosite (www.biosite.com) Triage system

    Cardiac, shortness of breath Proteins Biosite (www.biosite.com) Triage system

    Infectious disease Antibodies Bio-Rad Laboratories (www.bio-rad.com) Luminex +

    Infectious disease, Epstein-Barr Antibodies Zeus Scientific (www.zeusscientific.com) Luminex

    Infectious disease, FSME, Borrelia Antibodies Multimetrix (www.multimetrix.com ) Luminex

    Infectious disease, Herpes virus Antibodies Focus Diagnostics (www.focusdx.com) Luminex +

    Multiple immunoassays Proteins Randox (www.randox.com ) Evidence +

    Neurologic, Alzheimers disease Proteins Innogenetics (www.innogenetics.com) Luminex

    Typing, HLA Antibodies Tepnel (www.tepnel.com) Luminex +

    Genetic tests still dominate multiplexed diagnostics, but proteins are catching up. So far, predominantly antibodies have been detected in serum

    from patients suffering from allergies, autoimmune, or infectious diseases. Technically, the Luminex platform is the prevailing technology for

    multiplexed protein diagnostics (Source: Multiplexed Diagnostics 2008; www.SelectBiosciences.com)

    1412 M. Hartmann et al.

    http://www.biosite.com/http://www.biosite.com/http://www.biomerieux-usa.com/http://www.biomerieux-usa.com/http://www.invernessmedical.de/http://www.vbc-genomics.at/http://www.inovadx.com/http://www.immunetech.com/http://www.inbio.com/http://www.bioarrays.com/http://www.bmd-net.com/http://www.inovadx.com/http://www.zeusscientific.com/http://www.bio-rad.com/http://www.whatman.com/http://www.rulesbasedmedicine.com/http://www.biomerieux-diagnostics.com/http://www.biosite.com/http://www.biosite.com/http://www.bio-rad.com/http://www.zeusscientific.com/http://www.multimetrix.com/http://www.focusdx.com/http://www.randox.com/http://www.innogenetics.com/http://www.tepnel.com/http://www.selectbiosciences.com/http://www.selectbiosciences.com/http://www.tepnel.com/http://www.innogenetics.com/http://www.randox.com/http://www.focusdx.com/http://www.multimetrix.com/http://www.zeusscientific.com/http://www.bio-rad.com/http://www.biosite.com/http://www.biosite.com/http://www.biomerieux-diagnostics.com/http://www.rulesbasedmedicine.com/http://www.whatman.com/http://www.bio-rad.com/http://www.zeusscientific.com/http://www.inovadx.com/http://www.bmd-net.com/http://www.bioarrays.com/http://www.inbio.com/http://www.immunetech.com/http://www.inovadx.com/http://www.vbc-genomics.at/http://www.invernessmedical.de/http://www.biomerieux-usa.com/http://www.biomerieux-usa.com/http://www.biosite.com/http://www.biosite.com/
  • 7/31/2019 Protein Micro Arrays

    7/10

    with the same sensitivity and specificity as the diagnostic

    technologies that are currently used on a routine basis [41].

    Over the last ten years, antigen arrays with several hundred,

    and even up to several thousand, immobilized antigens have

    been used for detection of specific autoantibodies involved in

    autoimmune diseases. Joos et al. [42] used complex protein

    microarrays to detect up to 18 different rheumatic disease-

    specific autoantibodies in human sera, achieving sensitivitiesand specificities that were similar to established ELISA

    methods. This concept was further developed by implement-

    ing peptide antigens on a microarray, which allowed the

    detailed characterization of patients autoimmune statuses

    [43]. Protein microarrays are perfectly suited to such types of

    analysis, especially when the quantity of sample material is

    limited. Sharp et al. [44] suggested the application of

    autoantibody profiling to improve diagnosis and prediction

    of disease onset and severity. Protein microarrays are valu-

    able diagnostic tools for disease monitoring and therapy,

    because new clinical information can be gained from

    comprehensive autoantibody profiling, especially whencombined with other clinical parameters, e.g. cytokine levels.

    Arrays of viral and microbial antigens are the third type of

    antigen array. Mezzasoma et al. [45] successfully demon-

    strated the detection of antibodies directed against Toxo-

    plasma gondii, rubella virus, cytomegalovirus, and herpes

    simplex virus types 1 and 2 (ToRCH antigens) in serum

    samples. In a more comprehensive study, Waterboer et al.

    [46] used bead-based microarrays to analyze 756 sera for

    the presence of antibodies against 27 antigens derived from

    human papillomaviruses. Recombinant GST-tagged fusion

    proteins were bound to glutathione beads and incubated

    with patient sera. Bound antibodies were visualized using

    anti-human IgG-specific detection. The authors were able to

    correlate the protein array results with those from ELISA-

    based methods. However, multiplexed serology also en-

    abled them to effectively detect weak antibody responses.

    Gray et al. [47] used protein arrays containing immobilized

    malaria antigens to profile serum from malaria patients and

    found a correlation of the results with resistance to malaria.

    The authors analyzed combinations of reactivity to different

    antigens instead of individual reactivity and were able to

    group the patients according to their increased development

    of clinical immunity using hierarchical clustering. These

    studies demonstrated the potential of antigen microarrays,

    which can compete with ELISA tests in respect of sensitivity

    and robustness. Antigen microarrays are perfect tools for

    these types of application and support health personnel in

    diagnosis and prognosis.

    Antibody and reversed-phase microarrays

    Similarly to DNA microarrays used for mRNA expression

    analysis, protein-capture arrays are used in dual labeling

    approaches to investigate relative protein abundance in two

    differentially labeled samples. Haab et al. [30] demonstrat-

    ed the potential of such protein capture arrays by analyzing

    115 characterized antibody-antigen interactions. Capture

    antibodies were immobilized on poly-L-lysine-coated glass

    slides. Patient samples were labeled with Cy5 or Cy3 dye.

    Defined mixtures containing Cy5-labeled antigens at

    different concentrations were incubated with Cy3-labeledcontrol antigens. It was possible to achieve a correct linear

    relationship between antigen concentration and assay signal

    in about 20% of all interactions observed. Another 30% of

    the interactions reflected the antigen concentrations. Direct

    labeling approaches have already been commercialized by

    several companies (www.biochipnet.com). It must however

    be noted that sandwich immunoassays achieve higher spec-

    ificities and usually higher detection and assay sensitivities

    than direct labeling approaches.

    Focused protein microarrays such as miniaturized sand-

    wich immunoassays, either planar or bead-based, have

    evolved into tools that deliver many clinical data of highdiagnostic and prognostic value. For example, lyzed breast

    tumor biopsies samples were analyzed along with normal

    tissue for 14 relevant marker proteins from only 50 g

    protein using the bead-based Luminex xMAP system [48].

    The expression profile obtained for estrogen receptor and

    Her-2 (human epidermal growth factor receptor 2) exactly

    matched the results of the immunohistochemistry tests that

    are normally used for this type of application. Recently, the

    expression of 11 soluble receptors was analyzed in patient

    samples from 36 critically ill intensive care unit patients.

    Hierarchical clustering analysis allowed the scientists to

    group the patients into a sepsis and a trauma group [ 33].

    The field of inflammation holds huge potential for protein

    microarrays as diagnostic tools, especially in the field of

    sepsis, which is often hampered by the lack of quantitative

    IVD tests. Due to the complex nature of sepsis, many clinical

    parameters ought to be monitored in parallel. Protein micro-

    arrays are perfectly suited to fulfill this requirement [49].

    Reversed-phase microarrays also have enormous poten-

    tial as diagnostic tools, in particular in the identification of

    disease-specific biomarkers, which may be used as markers

    to initiate and monitor therapy. There are high expectations

    that biopsy analysis from individual patients may lead to

    therapies that are specifically tailored to individual require-

    ments [50].

    Commercial protein microarrays

    A variety of analytical protein microarrays have been

    developed for different platforms and are entering the

    diagnostic market (Table 1). The first planar and bead-

    based multiplex immunoassays have been cleared by the

    US FDA or have been CE-marked for use in the EU. These

    Protein microarrays for diagnostic assays 1413

    http://www.biochipnet.com/http://www.biochipnet.com/
  • 7/31/2019 Protein Micro Arrays

    8/10

    protein arrays are produced by the manufacturers whilst the

    assay and data analysis is performed by the customers. The

    AtheNA Multi-Lyte test system (Zeus Scientific, Raritan,

    USA) [51] and the BioPlex 2200 ANA screen (Bio-Rad,

    Hercules, USA) [52] are designed for autoimmune testing

    and are based on Luminexs xMAP technology. These

    multiplexed immunoassays can be used to screen samples

    for the presence of multiple autoantibodies involved inrheumatic diseases such as systemic lupus erythematosus

    (SLE), mixed connective tissue disease, Sjgrens syn-

    drome, scleroderma (systemic sclerosis), polymyositis, and

    CREST syndrome. The AtheNA Multi-Lyte test system

    uses intra-well calibration technology involving internal

    standards for analyzing patient serum characteristics. The

    measured assay signals are corrected to compensate assay

    drift and to allow patient-specific calibration. A positive

    result is achieved for each individual analyte test when the

    assay signal is above a specific upper threshold. The result

    is defined as negative when the assay signal is below a

    defined threshold. Signals between the lower and upperthresholds are marked as questionable for this specific

    analyte. The BioPlex 2200 ANA Screen system uses

    pattern-recognizing medical decision support software that

    associates test results with predefined patterns that have

    been correlated with autoimmune diseases. The k-nearest

    algorithm is used to check for greatest concordance with 11

    reference samples from a database containing data sets

    generated from more than 1,400 patient samples.

    CombiChip Autoimmune 1.0 from Whatman (Spring-

    field Mill, UK) is a planar microarray assay for autoim-

    mune diagnostics. This protein microarray uses 14 different

    autoantigens that are immobilized on nitrocellulose-coated

    slides within 16 identical subarrays. Using this autoimmune

    microarray, 16 patient samples per slide can be processed

    using multi-channel pipettes in the same way as conventional

    ELISA testing using microtiter plates. However, imaging and

    image analysis has to be done manually using common micro-

    array scanners and image analysis software. The CombiChip

    Autoimmune is CE-marked and sold in the EU; in the USA,

    the software is available for research use only.

    Randoxs Laboratories (Crumlin, UK) have developed

    Evidence, an automated biochip system enabling the

    analysis of miniaturized and parallelized immunoassays in

    a macroarray format containing 25 features and using

    chemiluminescence-based read out [53]. Several multiplexed

    immunoassays panels have been developed, including

    fertility, cardiac disease, tumors, cytokines and growth

    factors, cell adhesion molecules, thyroid function, and drug

    residues panels. The drugs abuse array has already received

    FDA clearance and other assays are currently being evaluated,

    thus making the Evidence biochip analyzer a pioneer in

    multiplexed immunoassays for clinical diagnostics.

    Problems and requirements

    Protein microarrays are well-established analytical tools in

    basic and applied research, which is a sign of the

    capabilities and power of these assay systems. Prior to

    their implementation in routine clinical diagnostics, protein

    microarray-based results have to demonstrate clinical

    relevance in the initiation or changing of therapy. Medicaldemand, combined with an overall cost reduction, must

    become the driving force behind protein arrays gaining a

    substantial share in the IVDs market. Although protein

    microarrays have huge diagnostic potential, they are

    nevertheless still far from being widely used in IVDs.

    Several regulatory hurdles have to be cleared, for example,

    the IVD tests have to fulfill the specifications of the 98/97/

    EC directive of the European Parliament and of the

    European Council of 22/12/98. In the USA, the Food and

    Drug Administration (FDA) decides on the approval of

    IVDs for human application. Before approval is granted,

    the tests have to provide valid results, and proof that theresults have a positive therapeutic outcome. In order to

    evaluate the reliability and reproducibility of DNA micro-

    arrays for gene expression analysis, the FDAs National

    Center for Toxicological Research (NCTR) set up the

    MicroArray Quality-Control (MAQC) project that involves

    both academic and commercial partners. (http://www.fda.

    gov/nctr/science/centers/toxicoinformatics/maqc ). In the

    first phase, more than 1,300 microarray experiments were

    performed on different platforms at different test sites with

    the aim of showing that a consensus in data analysis will

    make a considerable contribution to reproducible lists of

    differentially expressed genes [54]. In the next phase, the

    tests are further validated in terms of clinical applicability.

    Special focus is put on data analysis using different

    algorithms in order to obtain predictive signatures and

    classifiers. However, DNA analysis is unable to provide

    data of clinical relevance in the same way as protein

    analysis. Only proteins, and hence the proteome, are able to

    reflect the physiological state of a cell. DNA analysis

    enables the diagnosis of a persons genetic predisposition to

    a specific disease, but it will be not possible to predict the

    onset of disease. Although analytical protein microarrays

    are able to analyze fairly focused sets of analytes, a variety

    of problems have to be solved prior to the broad application

    of multiplexed protein microarrays on the IVD market. To

    ensure the high quality of microarrays, much more effort

    has to be made than with singleplex measurements. A

    variety of controls have already been implemented in

    protein microarrays, such as replicate spots, marker spots

    for orientation, negative control spots to check for

    unspecific binding, application of assay buffer to test for

    cross reactivity between capture molecules and detection

    1414 M. Hartmann et al.

    http://www.fda.gov/nctr/science/centers/toxicoinformatics/maqchttp://www.fda.gov/nctr/science/centers/toxicoinformatics/maqchttp://www.fda.gov/nctr/science/centers/toxicoinformatics/maqchttp://www.fda.gov/nctr/science/centers/toxicoinformatics/maqc
  • 7/31/2019 Protein Micro Arrays

    9/10

    antibodies, internal spot normalization [55, 56] and differ-

    ent normalization strategies [57]. However, quality control

    in terms of IVD testing means the routine analysis of

    reference sample material of low, medium and high

    concentration [58]. Only this will ensure that the test is

    effective at any analyte concentration and is able to detect

    an assay drift. At present, no information exists on the

    required complexity of such a reference sample and howstability can be guaranteed. The generation of defined

    mixtures of control analytes and the solubility of these

    molecules at high concentrations might cause problems that

    have not so far broadly investigated. Putting such controls

    in place will make it easier for the assay to be used for

    clinical analysis. However, additional issues will have to be

    addressed, for example the interpretation of assay results in

    cases when some of the controls fail. Can we regard the

    partial results gained from microarray with valid controls as

    sufficiently reliable or do these results have to be rejected,

    also? Another important issue is multiple subarrays within

    one slide. What happens if one of these subarrays fails?How can one tell whether the whole array is affected? How

    should failures within replicate spots be dealt with?

    Replicate spots are a compromise arising from the limited

    space within an array. Roches new microarray platform,

    Impact, employs up to 20 replicate spots [59] and bead-

    based systems analyze from fifty to one hundred beads

    [60]. In the case of a multiplexed diagnostic assay for the

    analysis of dozens of individual parameters being estab-

    lished and a few or even just one single test being changed,

    will a whole new approval for IVD be required?

    In addition, multiplexed assays generate huge data sets,

    which require appropriate data analysis. The question will be

    how to draw conclusions from patterns rather than looking

    into single interactions. The MicroArray Quality Control

    project (see above) was introduced to provide the necessary

    information for inter-platform and inter-laboratory concor-

    dance of DNA microarray experiments [54]. The BioPlex

    2200 ANA screen system uses a pattern-recognition algo-

    rithm for analysis of their multiplexed protein assays [52].

    However, there will be no general approach on how to apply

    such algorithms to generate diagnostically relevant data sets.

    Besides the regulatory aspects, social and ethical issues

    also need to be considered. Should a customer be allowed

    to order only part of the data generated with a multiplexed

    assay and therefore pay less than the price of the full

    multiplexed panel? Can the manufacturer or the performer

    sell only parts of the microarray results, for example by

    using different software settings? What happens to unre-

    quested data sets? What happens if the unrequested data

    sets are of diagnostic relevance that would lead to a

    different diagnosis? All these questions have to be carefully

    addressed and solved by the regulatory offices and by the

    diagnostic companies before protein microarrays are placed

    on the IVD market.

    Protein microarrays assays will have to be automated

    before entering the IVD market. Automation increases assay

    performance, robustness, and reliability of multiplexed

    assays. However, such automated multiplex platforms have

    to compete with the well-established clinical analyzers that

    currently dominate the diagnostic market. These systems caneasily increase throughput, e.g. measuring five parameters

    from the same sample in a sequential mode. Therefore, as

    long as sample material is not limited, or multiplexing does

    not exceed more than five parameters, the diagnostic

    companies are hesitant to make huge investments in order

    to change the assay format. In the field of autoimmune

    disease diagnostics, multiplexed assays are currently enter-

    ing the diagnostic market, and sets of tumor marker panels

    may in future also be applied to monitor therapy. It can be

    safely assumed that protein microarrays will find their place

    in the IVD market in areas in which sample volume is limited

    and where they deliver therapeutically relevant data sets.

    References

    1. Ekins RP (1998) Clin Chem 44:20152030

    2. Yalow RS, Berson SA (1960) J Clin Invest 39:11571175

    3. Saleem M (2008) Afr J Biotechnol 7:923925

    4. Feinberg JG (1961) Nature 192:985986

    5. Feinberg JG, Wheeler AW (1963) J Clin Pathol 16:282284

    6. Ekins RP (1989) J Pharm Biomed Anal 7:155168

    7. Hoheisel JD (2006) Nat Rev Genet 7:200210

    8. Plomin R, Schalkwyk LC (2007) Dev Sci 10:1923

    9. Ng JK, Liu WT (2006) Anal Bioanal Chem 386:427434

    10. Wang L, Luhm R, Lei M (2007) Adv Exp Med Biol 593:105116

    11. Gygi SP, Rochon Y, Franza BR, Aebersold R (1999) Mol Cell

    Biol 19:17201730

    12. MacBeath G (2002) Nat Genet 32:526532 Suppl

    13. Templin MF, Stoll D, Schwenk JM, Potz O, Kramer S, Joos TO

    (2003) Proteomics 3:21552166

    14. Zhu H, Bilgin M, Bangham R, Hall D, Casamayor A, Bertone P,

    Lan N, Jansen R, Bidlingmaier S, Houfek T, Mitchell T, Miller P,

    Dean RA, Gerstein M, Snyder M (2001) Science 293:21012105

    15. Kohler G, Milstein C (1975) Nature 256:495497

    16. Knappik A, Ge L, Honegger A, Pack P, Fischer M, Wellnhofer G,

    Hoess A, Wolle J, Pluckthun A, Virnekas B (2000) J Mol Biol

    296:5786

    17. Stoltenburg R, Reinemann C, Strehlitz B (2007) Biomol Eng

    24:381403

    18. Gunneriusson E, Nord K, Uhlen M, Nygren P (1999) Protein Eng

    12:873878

    19. Gunneriusson E, Samuelson P, Ringdahl J, Gronlund H, Nygren

    PA, Stahl S (1999) Appl Environ Microbiol 65:41344140

    20. Hober S, Uhlen M (2008) Curr Opin Biotechnol 19:3035

    21. Zhu H, Snyder M (2003) Curr Opin Chem Biol 7:55 63

    22. Angenendt P, Glokler J, Sobek J, Lehrach H, Cahill DJ (2003) J

    Chromatogr A 1009:97104

    23. Seurynck-Servoss SL, White AM, Baird CL, Rodland KD, Zangar

    RC (2007) Anal Biochem 371:105115

    Protein microarrays for diagnostic assays 1415

  • 7/31/2019 Protein Micro Arrays

    10/10

    24. Barbulovic-Nad I, Lucente M, Sun Y, Zhang M, Wheeler AR,

    Bussmann M (2006) Crit Rev Biotechnol 26:237259

    25. Templin MF, Stoll D, Bachmann J, Joos TO (2004) Comb Chem

    High Throughput Screen 7:223229

    26. Opalka D, Lachman CE, MacMullen SA, Jansen KU, Smith JF,

    Chirmule N, Esser MT (2003) Clin Diagn Lab Immunol 10:108

    115

    27. Yu H (1998) Anal Chim Acta 376:7781

    28. Hanash S (2003) Nature 422:226232

    29. Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P,Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP

    (1998) Nat Med 4:844847

    30. Haab BB, Dunham MJ, Brown PO (2001) Genome Biol 2:

    RESEARCH0004

    31. Kusnezow W, Banzon V, Schroder C, Schaal R, Hoheisel JD,

    Ruffer S, Luft P, Duschl A, Syagailo YV (2007) Proteomics

    7:17861799

    32. Schweitzer B, Roberts S, Grimwade B, Shao W, Wang M, Fu Q,

    Shu Q, Laroche I, Zhou Z, Tchernev VT, Christiansen J, Velleca

    M, Kingsmore SF (2002) Nat Biotechnol 20:359365

    33. Hsu HY, Wittemann S, Schneider EM, Weiss M, Joos TO (2008)

    Med Eng Phys doi:(10.1016/j.medengphy.2008.01.003) or PMID:

    18313970 (http://www.ncbi.nlm.nih.gov/pubmed/18313970)

    34. Baggio R, Carven GJ, Chiulli A, Palmer M, Stern LJ, Arenas JE

    (2005) J Biol Chem 280:4188419435. Usui-Aoki K, Shimada K, Nagano M, Kawai M, Koga H (2005)

    Proteomics 5:23962401

    36. Morais S, Carrascosa J, Mira D, Puchades R, Maquieira A (2007)

    Anal Chem 79:76287635

    37. Morais S, Tamarit-Lopez J, Carrascosa J, Puchades R, Maquieira

    A (2008) Anal Bioanal Chem 391:28372844

    38. Brown CR, Higgins KW, Frazer K, Schoelz LK, Dyminski JW,

    Marinkovich VA, Miller SP, Burd JF (1985) Clin Chem 31:1500

    1505

    39. Wiltshire S, OMalley S, Lambert J, Kukanskis K, Edgar D,

    Kingsmore SF, Schweitzer B (2000) Clin Chem 46:19901993

    40. Hiller R, Laffer S, Harwanegg C, Huber M, Schmidt WM,

    Twardosz A, Barletta B, Becker WM, Blaser K, Breiteneder H,

    Chapman M, Crameri R, Duchene M, Ferreira F, Fiebig H,

    Hoffmann-Sommergruber K, King TP, Kleber-Janke T, Kurup VP,

    Lehrer SB, Lidholm J, Muller U, Pini C, Reese G, Scheiner O,

    Scheynius A, Shen HD, Spitzauer S, Suck R, Swoboda I, Thomas

    W, Tinghino R, Van Hage-Hamsten M, Virtanen T, Kraft D,

    Muller MW, Valenta R (2002) FASEB J 16:414416

    41. Harwanegg C, Hiller R (2005) Clin Chem Lab Med 43:13211326

    42. Joos TO, Schrenk M, Hopfl P, Kroger K, Chowdhury U, Stoll D,

    Schorner D, Durr M, Herick K, Rupp S, Sohn K, Hammerle H

    (2000) Electrophoresis 21:26412650

    43. Robinson WH, DiGennaro C, Hueber W, Haab BB, Kamachi M,

    Dean EJ, Fournel S, Fong D, Genovese MC, de Vegvar HE,

    Skriner K, Hirschberg DL, Morris RI, Muller S, Pruijn GJ, van

    Venrooij WJ, Smolen JS, Brown PO, Steinman L, Utz PJ (2002)

    Nat Med 8:295301

    44. Sharp V, Utz PJ (2007) Nat Clin Pract Rheumatol 3:96103

    45. Mezzasoma L, Bacarese-Hamilton T, Di Cristina M, Rossi R,Bistoni F, Crisanti A (2002) Clin Chem 48:121130

    46. Waterboer T, Sehr P, Michael KM, Franceschi S, Nieland JD, Joos

    TO, Templin MF, Pawlita M (2005) Clin Chem 51:18451853

    47. Gray JC, Corran PH, Mangia E, Gaunt MW, Li Q, Tetteh KK,

    Polley SD, Conway DJ, Holder AA, Bacarese-Hamilton T, Riley

    EM, Crisanti A (2007) Clin Chem 53:12441253

    48. Schneiderhan-Marra N, Kirn A, Dttinger A, Templin M, Sauer

    G, Deissler H, Joos TO (2005) CGP 2:3742

    49. Heuer JG, Cummins DJ, Edmonds BT (2005) Expert Rev

    Proteomics 2:669680

    50. Speer R, Wulfkuhle J, Espina V, Aurajo R, Edmiston KH, Liotta

    LA, Petricoin EF (2007) Cancer Genomics Proteomics 4:157164

    51. Gilburd B, Abu-Shakra M, Shoenfeld Y, Giordano A, Bocci EB,

    delle Monache F, Gerli R (2004) Clin Dev Immunol 11:5356

    52. Shovman O, Gilburd B, Barzilai O, Shinar E, Larida B, Zandman-Goddard G, Binder SR, Shoenfeld Y (2005) Ann N Y Acad Sci

    1050:380388

    53. Fitzgerald SP, Lamont JV, McConnell RI, Benchikh el O (2005)

    Clin Chem 51:11651176

    54. Casciano DA, Woodcock J (2006) Nat Biotechnol 24:1103

    55. Hartmann M, Schrenk M, Dottinger A, Nagel S, Roeraade J, Joos

    TO, Templin MF (2008) Clin Chem 54:956963

    56. Olle EW, Sreekumar A, Warner RL, McClintock SD, Chinnaiyan

    AM, Bleavins MR, Anderson TD, Johnson KJ (2005) Mol Cell

    Proteomics 4:16641672

    57. Hamelinck D, Zhou H, Li L, Verweij C, Dillon D, Feng Z, Costa

    J, Haab BB (2005) Mol Cell Proteomics 4:773784

    58. Master SR, Bierl C, Kricka LJ (2006) Drug Discov Today

    11:10071011

    59. Hornauer H, Klause U, Mller H, Vieth F, Risse B (2004)

    Biospektrum 10:564565

    60. Fulton RJ, McDade RL, Smith PL, Kienker LJ, Kettman JR

    (1997) Clin Chem 43:17491756

    61. Templin MF, Stoll D, Schrenk M, Traub PC, Vohringer CF, Joos

    TO (2002) Trends Biotechnol 20:160166

    1416 M. Hartmann et al.

    http://dx.doi.org/10.1016/j.medengphy.2008.01.003http://www.ncbi.nlm.nih.gov/pubmed/18313970http://www.ncbi.nlm.nih.gov/pubmed/18313970http://dx.doi.org/10.1016/j.medengphy.2008.01.003